Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
J Med Biol Eng ; 41(5): 678-689, 2021.
Article in English | MEDLINE | ID: mdl-34483791

ABSTRACT

Purpose: In early 2020, the world is amid a significant pandemic due to the novel coronavirus disease outbreak, commonly called the COVID-19. Coronavirus is a lung infection disease caused by the Severe Acute Respiratory Syndrome Coronavirus 2 virus (SARS-CoV-2). Because of its high transmission rate, it is crucial to detect cases as soon as possible to effectively control the spread of this pandemic and treat patients in the early stages. RT-PCR-based kits are the current standard kits used for COVID-19 diagnosis, but these tests take much time despite their high precision. A faster automated diagnostic tool is required for the effective screening of COVID-19. Methods: In this study, a new semi-supervised feature learning technique is proposed to screen COVID-19 patients using chest CT scans. The model proposed in this study uses a three-step architecture, consisting of a convolutional autoencoder based unsupervised feature extractor, a multi-objective genetic algorithm (MOGA) based feature selector, and a Bagging Ensemble of support vector machines based binary classifier. The proposed architecture has been designed to provide precise and robust diagnostics for binary classification (COVID vs.nonCOVID). A dataset of 1252 COVID-19 CT scan images, collected from 60 patients, has been used to train and evaluate the model. Results: The best performing classifier within 127 ms per image achieved an accuracy of 98.79%, the precision of 98.47%, area under curve of 0.998, and an F1 score of 98.85% on 497 test images. The proposed model outperforms the current state of the art COVID-19 diagnostic techniques in terms of speed and accuracy. Conclusion: The experimental results prove the superiority of the proposed methodology in comparison to existing methods.The study also comprehensively compares various feature selection techniques and highlights the importance of feature selection in medical image data problems.

2.
Int J Oral Maxillofac Surg ; 50(8): 989-993, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33358522

ABSTRACT

Surgical practice during the coronavirus disease 2019 (COVID-19) pandemic has changed significantly, without supporting data. With increasing experience, a dichotomy of practice is emerging, challenging existing consensus guidelines. One such practice is elective tracheostomy. Here, we share our initial experience of head and neck cancer surgery in a COVID-19 tertiary care centre, emphasizing the evolved protocol of perioperative care when compared to pre-COVID-19 times. This was a prospective study of 21 patients with head and neck cancers undergoing surgery during the COVID-19 pandemic, compared to 193 historical controls. Changes in anaesthesia, surgery, and operating room practices were evaluated. A strict protocol was followed. One patient tested positive for COVID-19 preoperatively. There was a significant increase in pre-induction tracheostomies (28.6% vs 6.7%, P=0.005), median hospital stay (10 vs 7 days, P=0.001), and postponements of surgery (57.1% vs 27.5%, P=0.01), along with a significant decrease in flap reconstructions (33.3% vs 59.6%, P=0.03). There was no mortality and no difference in postoperative morbidity. No healthcare personnel became symptomatic for COVID-19 during this period. Tracheostomy is safe during the COVID-19 pandemic and rates have increased. Despite increased rescheduling of surgeries and longer hospital stays, definitive cancer care surgery has not been deferred and maximum patient and healthcare worker safety has been ensured.


Subject(s)
COVID-19 , Head and Neck Neoplasms , Head and Neck Neoplasms/surgery , Humans , Pandemics , Prospective Studies , SARS-CoV-2 , Tracheostomy
3.
3 Biotech ; 8(1): 17, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29270333

ABSTRACT

Out of 103 microsatellite markers used for studying the genetic diversity among local landraces of Luffa species, 56 were found polymorphic, including 38 gSSR and 18 eSSR, respectively. A total of 197 amplification products were obtained. The mean number of alleles per locus was 3.52. The PIC ranged from 0.037 to 0.986, while size of amplified product ranged from 105 to 500 bp. Cucumber-derived SSRs were amplified within L. acutangula (68%), L. aegyptiaca (61.16%), and L. hermaphrodita (60.2%), with an average of 63.12% cross-transferability. The Jaccard's coefficient ranged from 0.66 to 0.97, with an average of 0.81. High genetic variability was observed for node of 1st hermaphrodite flower (6.4-17), days to 1st hermaphrodite flower (38-52.1), days to 1st fruit harvest (43-65), number of fruit per cluster (1-5.9), fruit length (3.9-25 cm), fruit weight (18.4-175 g), number of fruit per plant (20-147.5), and yield per plant (2.2-4.7 kg). Two sub-populations were identified including 21 genotypes (sub-population I) and 06 genotypes (sub-population II), these two sub-populations showed 0.608-0.395% of the ancestral relationship to each other. This study provides information for future exploration, collection, and utilization of Luffa genotypes, as well as the polymorphic markers identified could be available for the study of landmarks in linkages, genomic structures, evolutionary ecology, and marker-assisted selection (MAS) in Luffa species.

4.
Nepal Med Coll J ; 16(2-4): 103-8, 2014 Dec.
Article in English | MEDLINE | ID: mdl-26930724

ABSTRACT

The aim of this study was to compare the level of glycated hemoglobin (HbA1c) in type 2 Diabetes Mellitus (DM) patients by two different methods namely Ion Exchange Chromatography and Affinity Binding Nycocard Reader. This is a cross-sectional study conducted on confirmed type 2 diabetes mellitus patients (n = 100) who visited Out Patients Department of the Universal College of Medical Sciences Teaching hospital, Bhairahawa, Nepal from November 2012 to March 2013. The diagnosis of diabetes mellitus was done on the basis of their fasting (164.46 ± 45.33 mg/dl) and random (187.93 ± 78.02 mg/dl) serum glucose level along with clinical history highly suggestive of type 2 DM. The HbA1c values of (7.8 ± 1.9%) and (8.0 ± 2.2%) were found in DM patients as estimated by those two different methods respectively. The highest frequency was observed in HbA1c > 8.0% indicating maximum cases were under very poor glycemic control. However, there were no significant differences observed in HbA1c value showing both methods are comparable in nature and can be used in lab for ease of estimation. The significant raised in HbA1c indicates complications associated with DM and monitoring of therapy become hard for those patients. Despite having standard reference method for HbA1c determination, the availability of report at the time of the patient visit can be made easy by using Nycocard Reader and Ion Exchange Chromatography techniques without any delay in communicating glycemic control, clinical decision-making and changes in treatment regimen.


Subject(s)
Diabetes Mellitus, Type 2/diagnosis , Glycated Hemoglobin/analysis , Adult , Aged , Aged, 80 and over , Chromatography, Ion Exchange , Cross-Sectional Studies , Diabetes Mellitus, Type 2/blood , Humans , Immunoassay/methods , Middle Aged , Young Adult
6.
J Pharmacol Exp Ther ; 299(3): 973-7, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11714885

ABSTRACT

A pandemic of obesity is contributing importantly to the prevalence of the metabolic syndrome characterized by hypertension, insulin resistance, and hyperlipidemia. In turn, the metabolic syndrome is contributing to vascular disease and the accelerating epidemic of chronic renal failure. Currently, pharmacological approaches to attenuate obesity and its cardiovascular/renal sequelae are limited. The purpose of this study was to determine the effects of 2-hydroxyestradiol, a metabolite of 17beta-estradiol with minimal estrogenic activity, on the development of obesity, the metabolic syndrome, and heart, vascular, and renal dysfunction in obese ZSF1 rats, a well-characterized genetic model of obesity and the metabolic syndrome with concomitant heart, vascular, and kidney disease. ZSF1 rats were treated, beginning at 12 weeks of age, for 26 weeks with vehicle or 2-hydroxyestradiol (10 microg/kg/h). At baseline and after 24 weeks of treatment, animals were placed in metabolic cages, and food intake, water intake, urine output, and urinary excretion of proteins and glucose were determined. Next, in fasting animals, plasma cholesterol was measured, an oral glucose tolerance test was conducted, and total glycated hemoglobin levels were determined. At the end of the study, animals were anesthetized and instrumented for assessment of heart performance, renal hemodynamics, and mesenteric vascular reactivity. 2-Hydroxyestradiol attenuated the development of obesity and improved endothelial function, decreased nephropathy, decreased the severity of diabetes, lowered arterial blood pressure, and reduced plasma cholesterol. 2-Hydroxyestradiol may be an important lead for the development of safe and effect drugs to attenuate obesity and its metabolic, vascular, and renal sequelae.


Subject(s)
Estradiol/analogs & derivatives , Estradiol/therapeutic use , Metabolic Diseases/prevention & control , Obesity/prevention & control , Renal Insufficiency/prevention & control , Animals , Male , Metabolic Diseases/complications , Obesity/complications , Rats , Renal Insufficiency/complications
7.
Arterioscler Thromb Vasc Biol ; 21(11): 1745-50, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11701460

ABSTRACT

Catechol-O-methyltransferase (COMT)-mediated methylation of 2-hydroxyestradiol (endogenous estradiol metabolite) to 2-methoxyestradiol (angiogenesis inhibitor) may be responsible for the antimitogenic effects of 2-hydroxyestradiol on vascular smooth muscle cells (VSMCs). Catecholamines are also substrates for COMT, and increased levels of catecholamines are associated with vasoocclusive disorders. We hypothesize that catecholamines may abrogate the vasoprotective effects of 2-hydroxyestradiol by competing for COMT and inhibiting 2-methoxyestradiol formation. To test this hypothesis, we investigated the antimitogenic effects of 0.001 to 0.1 micromol/L of 2-hydroxyestradiol on human aortic VSMC proliferation (cell number and DNA synthesis), collagen synthesis, and migration in the presence and absence of catecholamines. Norepinephrine, epinephrine, and isoproterenol concentration-dependently abrogated the inhibitory effects of 2-hydroxyestradiol on cell number, DNA synthesis, collagen synthesis, and cell migration. These modulatory/attenuating effects of catecholamines were not abrogated in the presence of the alpha- and beta-adrenergic receptor antagonists, phentolamine mesylate and propranolol, respectively. In contrast to 2-hydroxyestradiol, the antimitogenic effects of 2-methoxyestradiol (0.1 micromol/L) were not attenuated by isoproterenol (1 micromol/L) or quercetin (competitive inhibitor of COMT, 10 micromol/L). Norepinephrine, epinephrine, and isoproterenol concentration-dependently (10 to 500 micromol/L) inhibited the metabolism of 2-hydroxyestradiol (0.25 to 2 micromol/L) to 2-methoxyestradiol, and the potency of the catecholamines to reverse 2-hydroxyestradiol-induced inhibition of VSMC proliferation, collagen synthesis, and migration was correlated with their ability to inhibit 2-methoxyestradiol formation. Our findings suggest that catecholamines within the vasculature may abrogate the anti-vaso-occlusive effects of estradiol and 2-hydroxyestradiol by blocking 2-methoxyestradiol formation.


Subject(s)
Aorta/cytology , Epinephrine/pharmacology , Estradiol/analogs & derivatives , Estradiol/pharmacology , Isoproterenol/pharmacology , Muscle, Smooth, Vascular/metabolism , Norepinephrine/pharmacology , 2-Methoxyestradiol , Adrenergic alpha-Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cell Division/drug effects , Cell Movement/drug effects , Cells, Cultured , Collagen/biosynthesis , DNA/biosynthesis , Drug Antagonism , Estradiol/biosynthesis , Estradiol/metabolism , Female , Humans , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phentolamine/pharmacology , Propranolol/pharmacology
8.
J Appl Physiol (1985) ; 91(4): 1868-83, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11568174

ABSTRACT

17beta-estradiol (estradiol), the most abundant endogenous estrogen, affords cardiovascular protection. However, in a given cohort of postmenopausal women, estradiol replacement therapy provides cardiovascular protection in only a subset. The reasons for this variable action can only be understood once the mechanisms by which estradiol induces its cardiovascular protective effects are known. Because most biological effects of estradiol are mediated via estrogen receptors (ERs) and the heart and blood vessels contain both ER-alpha and ER-beta, the prevailing view is that ERs mediate estradiol-induced cardiovascular protection. However, recent findings that estradiol protects against vascular injury in arteries of mice lacking either ER-alpha or ER-beta seriously challenges this concept. Thus other non-ER mechanisms may be operative. Endogenous estradiol is enzymatically converted to several nonestrogenic metabolites, and some of these metabolites induce potent biological effects via ER-independent mechanisms. Therefore, it is conceivable that the cardiovascular protective effects of estradiol are mediated via its endogenous metabolites. On the basis of the evidence cited in this review, the cardiovascular protective effects of estradiol are both ER dependent and independent. The purpose of this article is to review the evidence regarding the cardiovascular protective effects of estradiol metabolites and to discuss the cellular, biochemical, and molecular mechanisms involved.


Subject(s)
Cardiovascular Diseases/physiopathology , Estradiol/physiology , Genome , Animals , Estradiol/genetics , Estradiol/metabolism , Female , Humans , Male , Sex Characteristics
9.
Am J Physiol Renal Physiol ; 281(4): F597-612, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11553506

ABSTRACT

Adenosine exerts physiologically significant receptor-mediated effects on renal function. For example, adenosine participates in the regulation of preglomerular and postglomerular vascular resistances, glomerular filtration rate, renin release, epithelial transport, intrarenal inflammation, and growth of mesangial and vascular smooth muscle cells. It is important, therefore, to understand the mechanisms that generate extracellular adenosine within the kidney. In addition to three "classic" pathways of adenosine biosynthesis, contemporary studies are revealing a novel mechanism for renal adenosine production termed the "extracellular cAMP-adenosine pathway." The extracellular cAMP-adenosine pathway is defined as the egress of cAMP from cells during activation of adenylyl cyclase, followed by the extracellular conversion of cAMP to adenosine by the serial actions of ecto-phosphodiesterase and ecto-5'-nucleotidase. This mechanism of extracellular adenosine production may provide hormonal control of adenosine levels in the cell-surface biophase in which adenosine receptors reside. Tight coupling of the site of adenosine production to the site of adenosine receptors would permit a low-capacity mechanism of adenosine biosynthesis to have a large impact on adenosine receptor activation. The purposes of this review are to summarize the physiological roles of adenosine in the kidney; to describe the classic pathways of renal adenosine biosynthesis; to review the evidence for the existence of the extracellular cAMP-adenosine pathway; and to describe possible physiological roles of the extracellular cAMP-adenosine pathway, with particular emphasis on the kidney.


Subject(s)
Adenosine/metabolism , Cyclic AMP/metabolism , Kidney/physiology , Animals , Extracellular Space/metabolism , Kidney/blood supply , Muscle, Smooth, Vascular/metabolism
10.
Hypertension ; 37(4): 1095-100, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11304509

ABSTRACT

Our previous studies show that cardiac fibroblasts express the extracellular "cAMP-adenosine pathway," that is, the generation of adenosine from extracelluar cAMP. The goal of this study was to assess whether activation of the cAMP-adenosine pathway by stimulation of endogenous cAMP synthesis regulates cardiac fibroblast growth. Cardiac fibroblasts in 3D cultures were used as the model system. Treatment of cardiac fibroblasts with forskolin, isoproterenol, or norepinephrine increased cAMP production and extracellular levels of adenosine, and these effects were prevented by inhibition of adenylyl cyclase (2',5'-dideoxyadenosine). Treatment with forskolin, isoproterenol, or norepinephrine for 24 hours inhibited DNA synthesis ((3)H-thymidine incorporation), and this effect was enhanced by combined inhibition of adenosine deaminase (erythro-9-[2-hydroxy-3-nonyl] adenine) plus adenosine kinase (iodotubercidin). Inhibition of adenylyl cyclase or adenosine receptors (1,3-dipropyl-8-p-sulfophenylxanthine or KF17837) prevented the effects of forskolin, isoproterenol, and norepinephrine on DNA synthesis. Forskolin also inhibited protein synthesis ((3)H-leucine incorporation) and cell proliferation, and these effects were blocked by adenosine receptor antagonism. Treatment of cardiac fibroblasts with norepinephrine for >48 hours but not <48 hours increased DNA synthesis, protein synthesis, and cell number. However, blockade of adenylyl cyclase or antagonism of adenosine receptors caused norepinephrine to induce proliferation in <48 hours. Our findings indicate that the endogenous cAMP-adenosine pathway regulates cardiac fibroblast growth.


Subject(s)
Adenosine/metabolism , Cyclic AMP/metabolism , Myocardium/cytology , Adenosine Deaminase Inhibitors , Adenosine Kinase/antagonists & inhibitors , Adenylyl Cyclase Inhibitors , Adrenergic alpha-Agonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Analysis of Variance , Animals , Cell Division/drug effects , Colforsin/pharmacology , DNA/biosynthesis , Fibroblasts/physiology , Isoproterenol/pharmacology , Male , Myocardium/metabolism , Norepinephrine/pharmacology , Protein Biosynthesis , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley
11.
Hypertension ; 37(2 Pt 2): 640-4, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230349

ABSTRACT

Estradiol inhibits endothelin-1 synthesis, an effect that may contribute to the cardiovascular protective effects of estradiol. Recent findings that estradiol inhibits neointima formation in mice lacking estrogen receptors suggests that the cardiovascular protective effects of estradiol may be mediated by means of an estrogen receptor-independent mechanism. Because 2-hydroxyestradiol and 2-methoxyestradiol, metabolites of estradiol with little/no affinity for estrogen receptors, are more potent than estradiol in inhibiting vascular smooth muscle cell growth, we investigated whether these metabolites also inhibit endothelin-1 synthesis by means of an receptor-independent mechanism. Treatment of porcine coronary artery endothelial cells for 4 to 24 hours with 0.001 to 1 micromol/L of estradiol, 2-hydroxyestradiol, or 2-methoxyestradiol concentration-dependently inhibited basal as well as serum-induced (2.5%), TNFalpha-induced (10 ng/mL), angiotensin II-induced (100 nmol/L), and thrombin-induced (4 U/mL) endothelin-1 synthesis. Estradiol, 2-hydroxyestradiol, and 2-methoxyestradiol also inhibited serum-induced mitogen-activated protein kinase activity. As compared with estradiol, its metabolites were more potent in inhibiting endothelin-1 secretion and mitogen activated protein kinase activity. The inhibitory effects of 2-hydroxyestradiol and 2-methoxyestradiol on endothelin-1 release and mitogen-activated protein kinase activity were not blocked by ICI182780 (50 micromol/L), an estrogen receptor antagonist. Our findings indicate that the estradiol metabolites 2-hydroxyestradiol and 2-methoxyestradiol potently inhibit endothelin-1 synthesis by means of an estrogen receptor-independent mechanism. This effect of estradiol metabolites may be mediated by inhibition of mitogen activated protein kinase activity and may contribute to the cardioprotective effects of estradiol.


Subject(s)
Endothelins/biosynthesis , Estradiol/analogs & derivatives , Estradiol/metabolism , Muscle, Smooth, Vascular/drug effects , 2-Methoxyestradiol , Animals , Cells, Cultured , Coronary Vessels/drug effects , Dose-Response Relationship, Drug , Endothelins/antagonists & inhibitors , Estradiol/pharmacology , Estrogen Receptor beta , Female , Fulvestrant , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/deficiency , Swine , Time Factors
12.
Hypertension ; 37(2 Pt 2): 645-50, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230350

ABSTRACT

Reduced nitric oxide synthesis by glomerular endothelial cells and increased proliferation of glomerular mesangial cells is associated with glomerular remodeling that leads to accelerated glomerulosclerosis. Estradiol induces nitric oxide synthesis and slows the progression of renal disease. Because the estradiol metabolites 2-hydroxyestradiol and 2-methoxyestradiol are more potent than estradiol in inhibiting growth of vascular smooth muscle cells, which are phenotypically similar to mesangial cells, we compared the effects of estradiol, 2-hydroxyestradiol, and 2-methoxyestradiol on growth of glomerular mesangial cells and on basal nitric oxide synthesis by glomerular endothelial cells. In human glomerular mesangial cells, estradiol and its metabolites concentration-dependently (1 nmol/L to 10 micromol/L) inhibited serum (2.5%)-induced DNA synthesis, cell proliferation, and collagen synthesis with the order of potency being 2-methoxyestradiol > 2-hydroxyestradiol > estradiol. ICI182780 (100 micromol/L, an estrogen receptor antagonist) blocked the growth inhibitory effects of estradiol but not 2-hydroxyestradiol or 2-methoxyestradiol. Treatment with estradiol, but not 2-hydroxyestradiol and 2-methoxyestradiol, induced nitric oxide synthesis (P<0.05, assayed by the formation of (3)H-L-citrulline from (3)H-L-arginine) in human glomerular endothelial cells, and these effects were blocked by ICI182780 and L-NMA (a nitric oxide synthesis inhibitor). In conclusion, estradiol may attenuate glomerulosclerosis by inducing nitric oxide synthesis via an estrogen receptor-dependent mechanism and by conversion to 2-hydroxyestradiol and 2-methoxyestradiol, which inhibit glomerular mesangial cell proliferation independent of estrogen receptors.


Subject(s)
Estradiol/analogs & derivatives , Estradiol/pharmacology , Kidney Glomerulus/drug effects , Nitric Oxide/metabolism , 2-Methoxyestradiol , Cell Division/drug effects , Cells, Cultured , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Estradiol/metabolism , Fulvestrant , Glomerular Mesangium/drug effects , Glomerular Mesangium/metabolism , Glomerulosclerosis, Focal Segmental/etiology , Humans , Kidney Glomerulus/metabolism , Nitric Oxide Synthase/metabolism , Progestins/pharmacology , Receptors, Estrogen/antagonists & inhibitors
13.
Hypertension ; 37(2 Pt 2): 658-62, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230352

ABSTRACT

Estradiol may be cardioprotective; however, the mechanisms involved remain unclear. Recent findings that estradiol attenuates neointima formation in estrogen receptor knockout mice suggest that the cardioprotective effects of estradiol may be mediated through estrogen receptor-independent mechanisms. Because 2-methoxyestradiol, an endogenous metabolite of estradiol with no affinity for estrogen receptors, is more potent than estradiol in inhibiting vascular smooth muscle cell growth, it is feasible that 2-methoxyestradiol mediates in part the cardioprotective effects of estradiol. To address this hypothesis, we examined the kinetics of 2-methoxyestradiol synthesis in vascular smooth muscle cells and endothelial cells. In human aortic smooth muscle cells, the V(max), K(m), and V(max)/K(m) ratio values for conversion of 2-hydroxyestradiol to 2-methoxyestradiol were 19+/-0.69 pmol. min(-1) per 10(6) cells, 0.52+/-0.085 micromol/L, and 44+/-4.9 pmol. min(-1). micromol/L per 10(6) cells, respectively. In human coronary artery vascular smooth muscle cells, the V(max), K(m), and V(max)/K(m) ratio values for conversion of 2-hydroxyestradiol to 2-methoxyestradiol were 16+/-0.59 pmol. min(-1) per 10(6) cells, 0.23+/-0.011 micromol/L, and 69+/-3.6 pmol. min(-1). micromol/L per 10(6) cells, respectively (all values significantly different compared with human aortic smooth muscle cells). Also, in human aortic versus coronary artery endothelial cells, the V(max) (33+/-0.24 versus 22+/-0.33 pmol. min(-1) per 10(6) cells, respectively), K(m) (0.20+/-0.010 versus 0.099+/-0.014 micromol/L, respectively), and V(max)/K:(m) (163+/-7.7 versus 243+/-41 pmol. min(-1). micromol/L per 10(6) cells, respectively) values were significantly different. Our results indicate that vascular smooth muscle and endothelial cells effectively metabolize 2-hydroxyestradiol to 2-methoxyestradiol. The lower K(m) and higher V(max)/K(m) ratio of human coronary versus aortic cells indicate a faster rate of local metabolism of 2-hydroxyestradiol to 2-methoxyestradiol in the coronary circulation at low levels of 2-hydroxyestradiol.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Estradiol/analogs & derivatives , Estradiol/biosynthesis , Estradiol/metabolism , Muscle, Smooth, Vascular/metabolism , 2-Methoxyestradiol , Animals , Aorta, Thoracic/metabolism , Cell Division/drug effects , Cells, Cultured , Epinephrine/pharmacology , Estradiol/pharmacology , Female , Humans , Isoproterenol/pharmacology , Kinetics , Methyltransferases/metabolism , Norepinephrine/pharmacology , Rats , Rats, Sprague-Dawley
14.
Hypertension ; 37(2 Pt 2): 716-21, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11230362

ABSTRACT

Adenosine inhibits growth of cardiac fibroblasts; however, the adenosine receptor subtype that mediates this antimitogenic effect remains undefined. Therefore, the goals of this study were to determine which adenosine receptor subtype mediates the antimitogenic effects of adenosine and to investigate the signal transduction mechanisms involved. In rat left ventricular cardiac fibroblasts, PDGF-BB (25 ng/mL) stimulated DNA synthesis ((3)H-thymidine incorporation), cellular proliferation (cell number), collagen synthesis ((3)H-proline incorporation), and MAP kinase activity. The adenosine receptor agonists 2-chloroadenosine and 5'-N-methylcarboxamidoadenosine, but not N(6)-cyclopentyladenosine, 4-aminobenzyl-5'-N-methylcarboxamidoadenosine, or CGS21680, inhibited the growth effects of PDGF-BB, an agonist profile consistent with an A(2B) receptor-mediated effect. The adenosine receptor antagonists KF17837 and 1,3-dipropyl-8-p-sulfophenylxanthine, but not 8-cyclopentyl-1,3-dipropylxanthine, blocked the growth-inhibitory effects of 2-chloroadenosine and 5'-N-methylcarboxamidoadenosine, an antagonist profile consistent with an A(2) receptor-mediated effect. Antisense, but not sense or scrambled, oligonucleotides to the A(2B) receptor stimulated basal and PDGF-induced DNA synthesis, cell proliferation, and collagen synthesis. Moreover, the growth-inhibitory effects of 2-chloroadenosine, 5'-N-methylcarboxamidoadenosine, and erythro-9-(2-hydroxy-3-nonyl) adenine plus iodotubericidin (inhibitors of adenosine deaminase and adenosine kinase, respectively) were abolished by antisense, but not scrambled or sense, oligonucleotides to the A(2B) receptor. Our findings strongly support the hypothesis that adenosine causes inhibition of CF growth by activating A(2B) receptors coupled to inhibition of MAP kinase activity. Thus, A(2B) receptors may play a critical role in regulating cardiac remodeling associated with CF proliferation. Pharmacologic or molecular biological activation of A(2B) receptors may prevent cardiac remodeling associated with hypertension, myocardial infarction, and myocardial reperfusion injury after ischemia.


Subject(s)
Adenosine/pharmacology , Mitogens/antagonists & inhibitors , Myocardium/metabolism , Receptors, Purinergic P1/metabolism , 2-Chloroadenosine/pharmacology , Adenosine/analogs & derivatives , Animals , Becaplermin , Cell Division , Cells, Cultured , Collagen/biosynthesis , Cyclic AMP/biosynthesis , DNA/biosynthesis , Enzyme Activation/drug effects , Fibroblasts/drug effects , Hypertension/physiopathology , Male , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinases/metabolism , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/physiopathology , Oligonucleotides, Antisense/pharmacology , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis , Purinergic P1 Receptor Agonists , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptor, Adenosine A2B , Receptors, Purinergic P1/genetics , Signal Transduction
15.
Am J Physiol Renal Physiol ; 280(3): F365-88, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11181399

ABSTRACT

A number of cellular and biochemical processes are involved in the pathophysiology of glomerular and vascular remodeling, leading to renal and vascular disorders, respectively. Although estradiol protects the renal and cardiovascular systems, the mechanisms involved remain unclear. In this review we provide a discussion of the cellular, biochemical, and molecular mechanisms by which estradiol may exert protective effects on the kidneys and vascular wall. In this regard, we consider the possible role of genomic vs. nongenomic mechanisms and estrogen receptor-dependent vs. estrogen receptor-independent mechanisms in mediating the protective effects of estradiol on the renal and cardiovascular systems.


Subject(s)
Cardiovascular System/drug effects , Estradiol/pharmacology , Kidney/drug effects , Protective Agents/pharmacology , Animals , Cardiovascular Physiological Phenomena , Cardiovascular System/cytology , Cardiovascular System/metabolism , Genome , Humans , Kidney/cytology , Kidney/metabolism
16.
Res Commun Mol Pathol Pharmacol ; 110(1-2): 107-16, 2001.
Article in English | MEDLINE | ID: mdl-12090349

ABSTRACT

Sepsis depletes intracellular stores of ATP and NAD+, leading to cellular energy failure. Liposome encapsulation improves intracellular delivery of bulky, charged molecules and substrates susceptible to extracellular enzyme degradation. We hypothesized that treatments with liposome encapsulated ATP or NAD+ would protect human endothelial cells exposed to endotoxin (LPS) and interferon-gamma (IFN-gamma) from energy failure. Liposomal ATP and NAD+ were prepared by a modification of the thin film method. Human endothelial cells were exposed to LPS 50 microg/ml and IFN-gamma 50 ng/ml for 72 hours, and liposomal ATP and NAD+ treatments were dosed at 0 and 24 hours. Energy state was determined by rate of mitochondrial respiration as measured by WST-1 assay. Mitochondrial respiration significantly decreased to 57% +/- 3 of control in LPS/IFN-gamma exposed cells after 72 hours. Liposomal ATP (200 microM) and NAD+ (100 microM) completely reversed this respiratory depression while empty liposomes, free ATP (200 microM). and free NAD+ (100 microM) did not. These results support the hypothesis that treatments with liposome encapsulated ATP or NAD+ protect human endothelial cells from energy failure in a cell culture model of sepsis and potentially may provide a novel therapy for use in clinical sepsis.


Subject(s)
Adenosine Triphosphate/administration & dosage , Endothelium, Vascular/drug effects , NAD/administration & dosage , Sepsis/drug therapy , Adenosine Triphosphate/therapeutic use , Cell Culture Techniques , Cell Respiration , Drug Delivery Systems , Endothelium, Vascular/metabolism , Energy Metabolism , Humans , Liposomes , Mitochondria, Muscle/metabolism , NAD/therapeutic use , Sepsis/metabolism
17.
Biochem Biophys Res Commun ; 278(1): 27-33, 2000 Nov 11.
Article in English | MEDLINE | ID: mdl-11071850

ABSTRACT

Estrogen receptors (ERs) are widely held to mediate the ability of 17 beta-estradiol (estradiol) to attenuate injury-induced proliferation of vascular smooth muscle cells (VSMCs) leading to vascular lesions. However, recent findings that estradiol prevents injury-induced vascular lesion formation in knock-out mice lacking either ER alpha or ER beta seriously challenge this concept. Here we report that the local metabolism of estradiol to methoxyestradiols, endogenous metabolites of estradiol with no affinity for ERs, is responsible for the ER-independent inhibitory effects of locally applied estradiol on rat VSMC growth. These finding imply that local vascular estradiol metabolism may be an important determinant of the cardiovascular protective effects of circulating estradiol. Thus, interindividual differences, either genetic or acquired, in the vascular metabolism of estradiol may define a given female's risk of cardiovascular disease and influence the cardiovascular benefit she receives from estradiol replacement therapy in the postmenopausal state. These findings also imply that nonfeminizing estradiol metabolites may confer cardiovascular protection in both women and men.


Subject(s)
Estradiol/analogs & derivatives , Estradiol/metabolism , Estradiol/physiology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , 2-Methoxyestradiol , Animals , Aorta/metabolism , Aorta, Thoracic/cytology , Catechol O-Methyltransferase/metabolism , Cell Count , Cell Division/drug effects , Cell Line , Cells, Cultured , Cytochrome P-450 Enzyme System/metabolism , Cytosol/metabolism , DNA/drug effects , Dose-Response Relationship, Drug , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Flavonoids/pharmacology , Fulvestrant , Humans , Inhibitory Concentration 50 , Luteolin , MAP Kinase Signaling System/drug effects , Male , Models, Biological , Protein Binding/drug effects , Quercetin/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/metabolism , Sex Factors , Time Factors , Tumor Cells, Cultured
18.
Hum Reprod Update ; 6(4): 332-50, 2000.
Article in English | MEDLINE | ID: mdl-10972521

ABSTRACT

The biology and physiology of the male as well as female reproductive system is hormonally regulated. Abnormalities in the dynamics of hormone production, metabolism and elimination, as well as their binding to certain target tissues, has been associated with pathophysiological conditions of the reproductive system. Although oestrogens are known to be one of the major hormone groups in regulating the reproductive function and the fertilization process, the cellular and biochemical mechanism or mechanism(s) via which oestrogens induce their effects are still not fully defined. Moreover, in a modern environment we are also exposed to a wide battery of environmental agents which are structurally similar to oestrogens, and termed 'environmental oestrogens'. Because environmental oestrogens have been shown to mimic some of the effects of oestradiol, it has been postulated that these exogenous chemicals may influence or interfere with the oestrogen-dependent reproductive processes, and may be associated with beneficial as well as deleterious effects on the reproductive system. In this regard, two classes of environmental oestrogens have been widely studied, i.e. phyto-oestrogens (plant-derived dietary oestrogens) and xeno-oestrogens (industrial chemicals, including polychlorinated biphenyls, DDT, TCDD, dioxins, etc.). The main focus of this review is to provide an overview on the cellular and biochemical mechanism(s) by which xeno-oestrogens and phyto-oestrogens influence the oestrogen-dependent reproductive functions and induce their deleterious or protective effects on the reproductive system.


Subject(s)
Cell Physiological Phenomena , Estrogens, Non-Steroidal/pharmacology , Estrogens/pharmacology , Isoflavones , Reproduction/drug effects , Xenobiotics/pharmacology , Animals , Estrogens/physiology , Humans , Phytoestrogens , Plant Preparations , Receptors, Estrogen/physiology , Reproduction/physiology
19.
Hypertension ; 36(3): 337-42, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10988261

ABSTRACT

The extracellular "cAMP-adenosine pathway" refers to the local production of adenosine mediated by cAMP egress into the extracellular space, conversion of cAMP to AMP by ectophosphodiesterase, and the metabolism of AMP to adenosine by ecto-5'-nucleotidase. The goal of this study was to assess whether the cAMP-adenosine pathway limits cardiac fibroblast growth. Studies were conducted in ventricular cardiac fibroblasts maintained in 3-dimensional cultures. Addition of exogenous cAMP to cardiac fibroblasts increased extracellular levels of AMP, adenosine, and inosine in a concentration-dependent and time-dependent manner. This effect was attenuated by blockade of total phosphodiesterase activity (3-isobutyl-1-methylxanthine), ectophosphodiesterase activity (high concentration of 1, 3-dipropyl-8-p-sulfophenylxanthine), or ecto-5'-nucleotidase (alpha, beta-methylene-adenosine-5'-diphosphate). Treatment with exogenous cAMP inhibited cell growth as assessed by DNA synthesis ((3)H-thymidine incorporation), cell proliferation (cell counts), and protein synthesis ((3)H-leucine incorporation). Antagonism of A(2) (KF17837) or A(1)/A(2) (low concentration of 1, 3-dipropyl-8-p-sulfophenylxanthine), but not A(1) (8-cyclopentyl-1, 3-dipropylxanthine), adenosine receptors blocked the growth-inhibitory effects of exogenous cAMP, but not the growth inhibitory effects of 8-bromo-cAMP (stable cAMP analogue). The growth-inhibitory effects of exogenous cAMP were enhanced by the combined inhibition of adenosine deaminase [erythro-9-(2-hydroxy-3-nonyl) adenine] and adenosine kinase (iodotubercidin). In conclusion, the extracellular cAMP-adenosine pathway exists in cardiac fibroblasts and attenuates cell growth. Pharmacological augmentation of this pathway could abate pathological cardiac remodeling in heart disease.


Subject(s)
Adenosine Monophosphate/metabolism , Adenosine/metabolism , Cyclic AMP/metabolism , Extracellular Space/metabolism , Fibroblasts/drug effects , Myocardium/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Analysis of Variance , Animals , Cell Division/drug effects , Cell Division/physiology , Cells, Cultured/drug effects , Cyclic AMP/pharmacology , Fibroblasts/cytology , Fibroblasts/metabolism , Inosine/metabolism , Male , Myocardium/cytology , Phosphodiesterase Inhibitors/pharmacology , Purinergic P1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Xanthines/pharmacology
20.
Arterioscler Thromb Vasc Biol ; 20(4): 964-72, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10764660

ABSTRACT

Some estrogenic compounds modify vascular smooth muscle cell (SMC) biology; however, whether such effects are mediated in part by estrogen receptors is unknown. The purpose of this study was to evaluate whether the actions of clinically used estrogens on human aortic SMC biology are mediated by estrogen receptors. We examined the effects of various clinically used estrogens in the presence and absence of ICI 182,780, an estrogen receptor antagonist, on cultured human aortic SMC DNA synthesis ([(3)H]thymidine incorporation), cellular proliferation (cell counting), cell migration (modified Boyden chamber), collagen synthesis ([(3)H]proline incorporation), and mitogen-activated protein kinase activity. FCS-induced DNA synthesis, cell proliferation, collagen synthesis, platelet-derived growth factor-induced SMC migration, and mitogen-activated protein kinase activity were significantly inhibited by physiological (10(-9) mol/L) concentrations of 17beta-estradiol and low concentrations (10(-8) to 10(-7) mol/L) of 17beta-estradiol, estradiol valerate, estradiol cypionate, and estradiol benzoate but not by estrone, estriol, 17alpha-estradiol, or estrone sulfate. The inhibitory effects of 17beta-estradiol and other inhibitory estrogens were completely reversed by 100 micromol/L ICI 182,780, and the rank-order potency of various estrogens to inhibit SMC biology matched their rank-order affinity for estrogen receptors. The inhibitory effects of estrogens on SMC biology are in part receptor-mediated. Because the cardioprotective effects of hormone replacement therapy are most likely mediated by modification of SMC biology, whether hormone replacement therapy protects a given postmenopausal woman against cardiovascular disease will depend partially on the affinity of the estrogen for estrogen receptors in vascular SMCs.


Subject(s)
Cell Division/drug effects , Enzyme Inhibitors/pharmacology , Estrogens/pharmacology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle, Smooth, Vascular/cytology , Aorta , Becaplermin , Cells, Cultured , DNA/biosynthesis , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estriol/pharmacology , Estrogen Antagonists/pharmacology , Estrone/analogs & derivatives , Estrone/pharmacology , Female , Fulvestrant , Humans , Muscle, Smooth, Vascular/metabolism , Platelet-Derived Growth Factor/pharmacology , Proto-Oncogene Proteins c-sis
SELECTION OF CITATIONS
SEARCH DETAIL
...